Host Defense and the Airway Epithelium: Frontline Responses That ...

11 downloads 3288 Views 730KB Size Report
Here we review mechanisms whereby airway epithelial cells recognize ..... such as the nucleotide oligomerization domain (NOD)-like receptors (NLRs), sense ...
SAGE-Hindawi Access to Research Journal of Pathogens Volume 2011, Article ID 249802, 16 pages doi:10.4061/2011/249802

Review Article Host Defense and the Airway Epithelium: Frontline Responses That Protect against Bacterial Invasion and Pneumonia Nicholas A. Eisele1, 2, 3 and Deborah M. Anderson1, 3 1 Department

of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO 65211, USA 3 The Laboratory for Infectious Disease Research, University of Missouri, Columbia, MO 65211, USA 2 Department

Correspondence should be addressed to Deborah M. Anderson, [email protected] Received 16 April 2011; Revised 18 July 2011; Accepted 21 July 2011 Academic Editor: Micahel Borchers Copyright © 2011 N. A. Eisele and D. M. Anderson. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Airway epithelial cells are the first line of defense against invading microbes, and they protect themselves through the production of carbohydrate and protein matrices concentrated with antimicrobial products. In addition, they act as sentinels, expressing pattern recognition receptors that become activated upon sensing bacterial products and stimulate downstream recruitment and activation of immune cells which clear invading microbes. Bacterial pathogens that successfully colonize the lungs must resist these mechanisms or inhibit their production, penetrate the epithelial barrier, and be prepared to resist a barrage of inflammation. Despite the enormous task at hand, relatively few virulence factors coordinate the battle with the epithelium while simultaneously providing resistance to inflammatory cells and causing injury to the lung. Here we review mechanisms whereby airway epithelial cells recognize pathogens and activate a program of antibacterial pathways to prevent colonization of the lung, along with a few examples of how bacteria disrupt these responses to cause pneumonia.

1. Introduction Host defense in the mammalian lung relies heavily on innate immune mechanisms that prevent invasion of pathogens. The airway epithelium is the front line defender of the lung which signals recruitment and activation of effector cells to kill invading pathogens and provides a physical barrier loaded with antibacterial compounds. Bacteria that successfully penetrate the epithelium must have the capability to evade these mechanisms, which typically means they avoid recognition and killing by both the effector cells of the innate immune system and the antimicrobial mechanisms in the epithelium. Pneumonia is a consequence of lung colonization, pathogen-induced injury to the epithelium, sustained activation of inflammation, and overactivation of tissue repair mechanisms. Furthermore, vascular leakage and edema are caused by these host responses, allowing the pathogen to gain access to the blood, where it may spread systemically and cause sepsis. Bronchopneumonia is characterized by focal areas of congestion of the parenchyma

by bacteria, inflammatory cells, and fibrin while lobar pneumonia is defined by a single area of congestion that takes up a larger portion of a lung lobe. Interstitial pneumonia involves congestion in the surrounding vasculature and is typically the result of overactive recruitment of inflammatory cells. In this paper, we will discuss how airway epithelial cells orchestrate innate immune responses in the lungs in order to limit invasion of bacterial pathogens, mediate tissue repair, and prevent pneumonia. The airway epithelium can be subdivided into bronchial and alveolar epithelial cells, which are polarized cells that share function in providing both a physical barrier and antimicrobial activity. The bronchial epithelial and goblet cells line the large airways, and these cells regulate ion exchange, mucin production, inflammation, and repair responses [1]. These cells form a physical barrier, connected by tight junctions, adherens junctions, and desmosomes that are relatively impermeable [2]. Similarly, the alveolar epithelium, composed of two distinct cell types, produces

2 antibacterial compounds such as surfactant, initiates and terminates inflammation, and regulates gas exchange to provide oxygen to the body. Resident alveolar macrophages and occasionally dendritic cells are also found in the alveolar epithelium and are key mediators of innate and adaptive immunity. Type I alveolar epithelial cells function primarily in facilitating gas exchange, but they also comprise a large portion of the impermeable barrier and can sense and respond to microbial products. Type II alveolar epithelial cells, also called type II pneumocytes, function as defenders of the airway through secreting antimicrobial products, sensing pathogenic invasion, and producing cytokines and chemokines that both activate and deactivate inflammation. In addition, type II cells can differentiate into type I cells and secrete repair enzymes upon damage to the epithelium. Mucins are continuously secreted by intraepithelial goblet cells and are composed of large glycoproteins that crosslink to form a structural barrier [1]. This property causes small and large particles, such as proteins and whole cells, to become trapped. Mucus contains a variety of antimicrobial compounds including IgA, collectins, and defensins which are regulated by the transcription factors NF-κB and Sp1 [2]. Microbes and other particulate material are pumped outward through the action of the mucociliated bronchial epithelium, a process that requires calcium transport, and serves as an environment for the activity of antimicrobial compounds [3]. Activation of NF-κB following signaling from the sentinel toll-like receptors (TLRs) induces epithelial cells to increase production of these compounds as well as proinflammatory cytokines which in turn also induce increased production of mucin. Together, these processes result in concentrated antimicrobial products that are broadly effective against invading microbes. Type II pneumocytes secrete surfactants on the apical side of the cell which are fatty acids that elicit similar functions as mucins. Four surfactant-associated proteins, SpA-D, are also produced by type II cells that function to agglutinate microbes to facilitate their clearance. Surfactant protein B has antimicrobial activity against bacteria by enhancing the phagocytic function of alveolar macrophages [4]. A number of antimicrobial products including βdefensins, lipocalin, and nitric oxide, as well as complement protein C3 and interferon are secreted into the surfactant layer (Table 1). Lipocalin chelates Fe+3 , limiting the access of bacteria to essential iron thereby stunting microbial growth [5]. β-defensins and nitric oxide provide direct killing of bacteria. β-defensins are small cationic antimicrobial peptides (CAMPs) with bactericidal activity and are attracted to the negative charge of the bacterial membrane. To maintain adequate concentrations of these peptides in the surfactant layer they are continuously secreted [6]. Type II epithelial cells also secrete repair enzymes, such as fibrinogen, on the basolateral face [7]. They quickly respond to changes in osmotic stress thereby able to sense nanomolar concentrations of bacterial pore-forming toxins. This allows cells to activate an inflammatory response long before the toxin is lethal to the cell [8]. Resident alveolar macrophages are phagocytic cells with distinct properties and initiate recruitment of inflammatory

Journal of Pathogens cells, such as neutrophils, as well as present antigen to cells of the adaptive immune system [9]. Uptake of pathogens by alveolar macrophages can be stimulated through antibody or complement opsonization, both of which are produced by airway epithelial cells and are also present in the blood. Recognition of intracellular or extracellular bacteria by pattern recognition receptors (PRRs) expressed by alveolar epithelial cells and macrophages stimulates an increase in the production of antimicrobial compounds, complement, cytokines, and chemokines which recruit effector cells such as neutrophils, monocytes, and dendritic cells, as well as T and B cells (Table 1) [10]. Inflammatory monocytes and neutrophils, however, must not reside in the lung long term because they produce and secrete cytotoxic molecules that will injure the delicate type I cells. Instead, polarized secretion of cytokines and chemokines by type I and II cells promotes recruitment, adherence, and transepithelial migration of inflammatory cells to fight infection, which is subsequently downregulated to limit their damage to the epithelium [11]. Bacterial pathogens penetrate the epithelium either through its disruption or by directly invading the airway epithelial cells. Disruption can be achieved by the induction of apoptosis or the use of bacterial exotoxins that directly lyse cells [12]. The use of toxins to promote penetration of epithelial barriers triggers injury responses that activate inflammation independent of pathogen recognition. In many cases, pathogens delay or prevent repair of the epithelium and these injury responses exacerbate lung congestion and accelerate disease [13]. In addition, some opportunistic pathogens derive help in crossing the epithelium through coinfection with viruses that impair mucociliary function, thereby allowing disruption of the epithelial barrier. Many bacterial pathogens have obligate or facultative intracellular life cycles enabling them to invade the epithelium, survive, and replicate in multiple environments. Bronchial and alveolar epithelial cells are not naturally phagocytic. Rather, some bacteria carry virulence factors that promote their entry into these cells. Once inside the epithelial cells, pathogens encode mechanisms that subvert normal trafficking pathways such that they may replicate in a membrane bound compartment or the cytoplasm. In order to replicate extracellularly, organisms must escape the cell which typically involves lysis caused by bacterial poreforming toxins, induction of apoptosis, or may simply be the result of massive replication.

2. Pathogen Detection Detection of conserved structural motifs, termed pathogen associated molecular patterns (PAMPs), is achieved by the expression of surface receptors, both on the cell surface and in endosomes. Detection of an invading bacterium through a particular PRR results in the production of proinflammatory cytokines and chemokines that recruit and activate immune effector cells, such as granulocytes and T cells, to the site of infection. In addition to mediating a localized inflammatory

Journal of Pathogens

3 Table 1: Immunomodulatory roles of type II alveolar epithelial cells.

Role

Component

Function

Ref

Complement Cathelicidin β-defensins Immunoglobulin Lipocalin 2 Lysozyme Nitric oxide Surfactant

Membrane disruption, opsonization, and inflammation Membrane disruption Membrane disruption Complement-mediated lysis, agglutination, and opsonization Iron sequestration Membrane disruption Membrane disruption Agglutination (SpA, SpD), membrane disruption (SpB)

[6] [6] [6] [6] [6] [6] [5] [6]

TLR2

Recognition of lipoproteins, lipoteichoic acid, and peptidoglycan

[6]

TLR4 IL-2R TNF-R1 MHC-I MHC-II IL-1α IL-1β IL-4 IL-6 IL-8 GRO-α ENA-78 MIP-2 TNF-α GM-CSF RANTES MCP-1 IFN-α/β IFN-γ

Recognition of lipopolysaccharide IL-2 receptor TNFα receptor Antigen presentation to CD8 T cells Antigen presentation to CD4 T cells Prostaglandin production, induces TNFα Prostaglandin production, induces TNFα Th2 Polarization, immunoglobulin production T cell recruitment, B cell differentiation Neutrophil chemotaxis Neutrophil chemotaxis Neutrophil chemotaxis Neutrophil chemotaxis Vasodilation, neutrophil activation Granulocyte and monocyte differentiation Monocyte and T cell recruitment Monocyte and T cell recruitment MHC-I Expression, NK cell activation Th1 Polarization, macrophage activation

[6] [5] [5] [6] [6] [6] [6] [5] [6] [6] [6] [6] [6] [6] [5] [6] [6] [6] [5]

Antimicrobial

Inflammation

Receptors

Cytokines and chemokines

response, soluble PRRs and antimicrobial peptides can also directly mediate killing of invading organisms through disruption of the cell membrane, resulting in osmotic lysis of bacteria [14]. Toll-like receptors (TLR) are transmembrane proteins that form a major family of PRRs and are ubiquitously expressed. TLRs form homo- and heterodimers with other TLRs or accessory proteins that are together responsible for the recognition of a variety of PAMPs including bacterial lipoprotein, lipopolysaccharide (LPS), peptidoglycan, flagellin, RNA, and nonmethylated CpG DNA. Different complexes are thought to mediate specific signal transduction pathways allowing for an increased repertoire of downstream responses. These proteins signal both innate, and adaptive immune responses and their collective action is essential for immune defense against bacterial pathogens. TLR2, 4, and 5 detect most species of bacteria and each of these signal through the common adaptor protein MyD88, located on the cytoplasmic face of the plasma membrane.

Signal transduction via phosphorylation cascade activates the nuclear translocation of NF-κB, which leads to the production of proinflammatory cytokines and chemokines. In addition, MyD88-independent signaling can also occur, leading to the expression of type I interferons (IFN-I) which also activate expression of proinflammatory cytokines and chemokines [15]. Bacterial LPS is composed of lipid A, core polysaccharide, and O-antigen. Lipid A composition varies between bacterial species and between cells of the same species and plays an important role in the pathogen’s ability to colonize the host lung. Recognition of lipid A is achieved by the delivery of monomeric LPS micelles to CD14 by soluble LPS-binding protein (LBP) [16]. TLR4, in complex with the accessory protein MD2, then associates with CD14 forming the TLR4MD2-CD14 LPS receptor complex. Upon complex formation, the cytoplasmic tail of TLR4 is able to associate with MyD88 leading to NF-κB activation. Alternatively, TLR4 can also signal through the adaptor TRIF/TRAM leading

4 to the activation of the transcription factor IRF-3 and the subsequent expression of type I interferon [17, 18]. Both the MyD88-dependent and independent pathways are important for host defense in the lungs, as mice deficient in these processes are more susceptible to pulmonary infection by many, but not all, bacterial species. Moreover, each pathway plays a distinct role against specific pathogens. TLR4 signaling can be initiated either on the cell surface or from the cytoplasm where the receptor remains associated with pathogen containing endosomes. In addition, TLR4 signaling induces crosstalk with other PRRs. For example, TLR4 activation upregulates surface expression of TLR2, enhancing the capacity of cells to be activated in response to PAMPs [19]. TLR2 primarily responds to Gram-positive bacteria through the detection of lipoteichoic acid and peptidoglycan from the apical surface of airway epithelial cells [20]. TLR2 has several coreceptors, including CD14 and CD36, which likely lead to specific activation patterns. In addition, gangliosides, some of which function as receptors for pathogen invasion, can also act as coreceptors for TLR2 and alter its ability to respond to PAMPs. Like TLR4, TLR2 signaling can be MyD88-dependent or independent, resulting in NF-κB or IRF-3 activation and can be activated from the cell surface or from intracellular compartments [21]. In addition to the toll-like receptors, other PRRs such as the nucleotide oligomerization domain (NOD)-like receptors (NLRs), sense intracellular pathogens through the detection of peptidoglycan and bacterial DNA in the host cell cytoplasm. Retinoid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) are RNA helicases that recognize microbial RNA and methylated DNA in the cytoplasm and activate IRF-3 [22]. In addition, DNA activator of interferon regulatory factors (DAI) is a major cytosolic DNA sensor that also leads to the activation of IRF-3 [23–25]. Thus, bacterial DNA is a potent inducer of inflammation through its effect on IFN-I gene expression from epithelial cells as well as alveolar macrophages. TLR2, 4, and 5 are the primary sensors of bacteria, and crosstalk occurs between other toll-like receptors that respond to viral infection. For example, viral activation of TLR3 not only stimulates production of proinflammatory cytokines in type II cells but also leads to upregulation of TLR2 and some, but not all, of its coreceptors allowing for enhanced detection of bacteria [26]. In contrast, TLR5 is downregulated following TLR3 activation thereby decreasing the ability to respond to bacterial flagellin, leaving an opportunity for bacterial coinfection. In addition to impacting PAMP recognition, viral infection may also assist in the deterioration of the airway epithelium or in modulating immune responses, leading to increased susceptibility to secondary bacterial pneumonia caused by opportunistic pathogens [27]. Many viruses destroy mucus producing or ciliated cells. Alternatively, viral infection may activate cell death pathways that then influence the ability of epithelial cells to respond to secondary infection. For example, following influenza virus infection, the host recruits monocytes to the airways which contribute proapoptotic signals to type I epithelial cells [28]. The resulting decay of the alveolar barrier can be exploited by

Journal of Pathogens opportunistic pathogens now able to invade and grow within the air spaces. Furthermore, influenza infection also depletes the host of effector cells such as monocytes, macrophages, and natural killer (NK) cells [28, 29].

3. Pathogen Clearance and Resolution of Inflammation Cytokines and chemokines produced by airway epithelial cells rapidly stimulate recruitment and activation of neutrophils, eosinophils, monocytes, dendritic cells (DCs), and NK cells which are capable of destroying invading bacteria [30–33]. By 6 or 7 days postinfection, T cells are also recruited by RANTES or IP-10 production (for Th1 cells) and IL-1β (for Th2 cells). Subsequently, airway epithelial cells receive signals from recruited inflammatory cells to increase production of defense mechanisms. For example, elastase is a bactericidal serine protease stored in granuoles and is secreted by activated neutrophils in response to infection [34]. Following release of the granules, bronchial epithelial cells respond to elastase by upregulating the expression of βdefensins [35]. Because inflammatory cell activity in the airway is likely to cause tissue damage and congestion that interferes with lung function, epithelial cells also mediate downregulation of inflammatory responses following bacterial clearance in order to protect the lung from unnecessary tissue damage. Production of glucocorticoids and other lipids, as well as anti-inflammatory cytokines, such as IL-10 and TGF-β, by airway epithelial cells help to downregulate inflammation [36]. Lysophosphatidic acid (LPA) is a mediator produced by epithelial cells that regulates expression of pro- and antiinflammatory cytokines and chemokines. This phospholipid acts by blocking the binding of interferon response factor1 (IRF-1) to DNA, thereby reducing its ability to stimulate expression of downstream proinflammatory genes. LPA can also induce expression of lipid mediators of inflammation as well as repair enzymes. Costimulatory signals influence downstream responses, for example, IFN-γ and TNF-α costimulation of bronchial epithelial cells results in LPAmediated downregulation of the neutrophil chemokine CCL5/RANTES. As IFN-γ and TNF-α are induced by pathogens and accumulate in the bronchus, LPA production by epithelial cells prevents long-term recruitment of neutrophils to prevent unnecessary damage to the lung.

4. Bacterial Pneumonia When these defense mechanisms fail to prevent bacterial infection, pneumonia rapidly develops. Bacterial pneumonia can be subdivided into community acquired and hospital acquired. The most common community and hospital acquired pneumonia is caused by Streptococcus pneumoniae, an extracellular, opportunistic pathogen whose virulence is primarily derived from the production of a capsule which allows the organism to adhere to mucosal tissues and evade the innate immune responses of the lung, including surfactants, complement, and phagocytosis [37]. Others,

Journal of Pathogens such as Klebsiella pneumoniae and Acinetobacter baumannii, primarily cause hospital acquired infections using similar strategies [38, 39]. In contrast, community acquired pneumonia can be caused by other pathogens that target the lung as a primary replicative niche using multiple virulence strategies. For example, Francisella tularensis, Staphylococcus aureus, and Yersinia pestis cause lung injury as extracellular pathogens through the production of secreted toxins, but employ an intracellular life cycle that relies on entirely different virulence mechanisms to invade the epithelium, evade innate immune detection, and establish a successful infection. These pathogens have in common the capacity to produce numerous modulators of detection, as well as virulence factors that serve multiple roles during infection. However, the mechanisms whereby these pathogens evade the onslaught of detection and destruction initiated by airway epithelial cells are very different. Because of their abilities to disable multiple levels of innate immunity in the lungs, we will discuss these three pathogens in greater detail. 4.1. Francisella tularensis. Francisella tularensis infection through tick transmission, wound infection, or inhalation results in a number of disease manifestations in humans such as oculoglandular, oropharyngeal, gastrointestinal, typhoidal, and pneumonic tularemia [40]. Several subspecies have been identified and have varying degrees of pathogenicity, with F. tularensis subsp. tularensis (type A strain) being the most lethal, and F. tularensis subsp. holarctica (type B strain) being less pathogenic. F. tularensis subsp. holarctica is the parent strain of the attenuated Live Vaccine Strain (LVS) which was established by serial passage in the laboratory and was used as a vaccine for many years. A third subspecies, F. tularensis subsp. novicida, rarely causes disease in humans but is highly virulent in mice and is also routinely used as a model system for F. tularensis research [41]. Important mechanisms of virulence and immunity have been elucidated in the attenuated Francisella strains, yet significant differences exist in the ability of the human pathogens to control inflammatory responses and cause disease. Pneumonic disease develops as a sequela of systemic infection, or as a result of inhaling bacteria and is characterized as a lobar pneumonia [40]. Symptoms are typically nonspecific and include fever, headache, and muscle aches, but patients can also present more severe indicators such as chest pain, bloody sputum, and dyspnea [42]. If left untreated, mortality rates reach 30–60%. Francisella is a facultative intracellular pathogen that targets macrophages and epithelial cells, and this interaction is essential for virulence. Bacteria interact with a multitude of receptors and are taken up by alveolar macrophages following opsonization in a process termed “looping phagocytosis” [43–45]. Nascent Francisella containing phagosomes do not mature into phagolysosomes but instead are lysed, allowing the bacteria to escape to the cytoplasm and replicate [46–48]. Escape from the phagosome and intracellular replication is dependent on the Francisella pathogenicity island (FPI) which encodes a Type VI secretion system [49, 50]. Expression of

5 FPI genes is dependent on the global virulence regulator MglA [51]. Organisms eventually reside in autophagous vacuoles termed Francisella containing vacuoles (FCV) [48]. High level intracellular replication is thought to ultimately lead to macrophage lysis and spread of extracellular bacteria [52]. The presence of IFN-γ significantly increases the resistance of mice to Francisella infection [53, 54]. IFN-γ signaling through the IFN-γ receptor stimulates macrophages to upregulate bactericidal effector function against all three Francisella strains [55]. In murine and human macrophages, IFN-γ inhibits the intracellular growth of F. novicida and LVS in an iNOS-independent (inducible nitric oxide synthase) manner, likely by preventing bacteria from escaping the phagosome thereby promoting lysosomal fusion [56–58]. In contrast, F. tularensis phagosomal escape is not inhibited by IFN-γ, and growth is instead restricted in the cytoplasm [59]. The outer membrane protein OmpC of F. novicida inhibits IFN-γ induced STAT1 phosphorylation in macrophages [60]. Although OmpC is conserved in the other Francisella spp., further studies are needed to determine if OmpC also blunts IFN-γ signaling for the human pathogens. Similar to other Gram-negative bacteria, Francisella has an LPS structure that contains lipid A, core, and O-antigen domains that are important to the pathogenesis of the organism [16, 61]. Francisella lipid A does not elicit an inflammatory response through TLR4 due to a lack of LBP binding [62–65]. Structural comparisons of Francisella lipid A to those that are highly proinflammatory, such as found in E. coli, indicate that its dephosphorylated glucosamine backbone and tetra-acylation may not be recognized by LBP, thereby preventing TLR4 signaling [66–70]. Artificial stimulation of TLR4 pathways before and after F. novicida infection, or use of a strain altered in lipid A structure (flmF2, flmK, and lpxF) induces an inflammatory response that lowers bacterial burden in the lung and increases the survival of infected mice, illustrating the importance of avoiding LPSdependent TLR4 activation for pathogenesis [70–72]. In addition, intact Francisella LPS core has been shown to be important in regulating cytotoxicity towards macrophages [73]. When core components are mutated, the bacteria are attenuated and hypercytotoxicity towards J774.A1 macrophages is observed and that results in attenuated virulence. Interestingly, core mutants are able to invade macrophages treated with cytochalasin-D, indicating that uptake can be independent of phagocytosis and suggesting the existence of a surface located receptor. Moreover, the enhanced cytotoxicity of core mutants is caused by intracellular bacteria and is independent of TLR4. LPS O-antigen polysaccharide also contributes to Francisella pathogenicity by modulation of complement C3 activity. C3 deposition occurs on the surface of all virulent Francisella strains, but surface located C3b is converted to C3bi [74]. While C3b leads to cell lysis and opsonization, C3bi is only opsonizing and greatly enhances phagocytic uptake [43, 45, 75]. C4b and factor H deposition occurs normally on the surface of bacteria, but the C5b-C9 membrane attack complex (MAC) does not form, resulting in resistance to complement-mediated lysis [76]. Altered

6 O-polysaccharide structures (as occurs in the LVS strain), renders the bacteria sensitive to complement through the C3b pathway and results in attenuation of virulence [74]. O-antigen from F. tularensis and LVS forms a capsular polysaccharide around the organism [77]. Loss of this capsule results in increased serum killing, reduced intracellular replication and hyper cytotoxicity toward macrophages [77, 78]. Conversely, F. novicida does not produce an O-antigen polysaccharide capsule, yet it is still resistant to complementmediated lysis suggesting that distinct mechanisms of complement resistance may exist in this related strain [74, 77]. Passive transfer of serum raised against bacteria lacking the O-antigen, or immunization of mice with such a strain, does not protect mice from challenge by wild type bacteria, indicating that the O-antigen is also important for the development of humoral immunity [79, 80]. In addition to resisting the downstream effects of complement deposition, Francisella has also acquired mechanisms to resist human β-defensins (hBD) [81]. hBD-1 and hBD-2 show minimal to moderate bactericidal activity, respectively, against LVS and F. novicida, but only at artificially high concentrations. Conversely, hBD-3, which has potent antimicrobial activity towards many microorganisms, kills Francisella effectively [82, 83]. hBD-1 is constitutively expressed by lung type II epithelial cells while hBD-2 and hBD-3 expression is inducible [84, 85]. Consequently, to evade hBD-3, Francisella suppresses its expression in type II cells [81]. The mechanism of this suppression and the bacterial virulence factor(s) involved remain unknown. Although Francisella has evolved a hypoinflammatory cell surface, mammalian hosts have coevolved methods to detect the organism through alternative mechanisms. To this end, it has been known for some time that TLR2 activation and MyD88-dependent and independent signaling as well as induction of the inflammasome can result in effective clearance of Francisella in mouse models, and crosstalk between these pathways is critical [62, 86–91]. TLR2 recognition of Francisella lipoproteins likely occurs in the phagosome and results in protection of mice from lethal infection [62, 87, 92, 93]. Cell based assays indicate that LVS decreases TLR2induced inflammation by activating phosphatidylinositol 3-kinase (PI3K) and upregulating MAPK phosphatase-1 (MKP-1), resulting in suppressed proinflammatory cytokine production from infected macrophages [94]. Whether this also occurs in airway epithelial cells has not yet been determined. Until recently, in fact, relatively little attention has been given to the role of alveolar epithelial cells in Francisella pathogenesis, but bacteria are able to invade these cells in vitro and in vivo [95]. To adhere to type II cells, LVS expresses FsaP (F. tularensis surface associated protein), which promotes tight association to epithelial cells in vitro and may also use type IV pili [96, 97]. Similar to macrophages, invasion into alveolar epithelial cells is dependent on a preformed bacterial surface structure, as live and dead bacteria are internalized in a manner indistinguishable from one another. Invasion requires cells to be competent in cytoskeleton rearrangement, as inhibiting microfilament and microtubule activity abrogates internalization suggesting that it may

Journal of Pathogens enter through endocytosis. Internalized bacteria initially colocalize with the early endosomal marker EEA1, then later with the lysosomal marker LAMP-1. Bacteria then escape this compartment and replicate in the cytoplasm [98]. Interestingly, type II pneumocytes are stimulated to produce proinflammatory cytokines in vitro by F. tularensis, and thus, it is unclear whether airway epithelial cells sense and appropriately respond to invading bacteria [99]. Upon pulmonary challenge with Francisella, proinflammatory chemokine production by type II pneumocytes, as well as induction of matrix metalloproteinase-9 (MMP9) and subsequent breakdown of the extracellular matrix, recruits neutrophils to the sites of infection [99, 100]. However, recent evidence suggests that during transendothelial migration, neutrophils acquire a depressed inflammatory phenotype that prevents exogenous activation [101]. Artificial depletion or recruitment of neutrophils during infection has little impact on the outcome of disease, indicating that bacteria are either resistant to neutrophil effector function, other cell types are able to control the infection, or both [102]. Subsequent analysis has shown that LVS inhibits NADPH oxidase assembly, supporting the hypothesis that bacteria are resistant to neutrophil function [103, 104]. Multiple virulence genes are required for LVS to inhibit oxidative burst in human neutrophils, including a number of acid phosphatases as well as pyrimidine biosynthesis genes [105–107]. 4.2. Staphylococcus aureus. Staphylococcus aureus is a Grampositive, opportunistic human pathogen, a commensal of the skin that colonizes an estimated 30% of the population and is a leading cause of hospital acquired infection. More recently, in addition to antibiotic resistant strains, more invasive isolates have emerged due to numerous genes acquired by this constantly evolving pathogen [108, 109]. Staphylococcal endocarditis, pneumonia, and sepsis now pose significant threats to both healthy and immune compromised individuals. These strains, known as community associated methicillin resistant S. aureus (CA-MRSA), now predominate in the human population worldwide and are no longer limited to nosocomial infections. Innate immune recognition of S. aureus is largely achieved through TLR2-dependent recognition of lipoteichoic acids, lipoproteins, and peptidoglycan, both from the cell surface and on endosomes of antigen presenting cells and type II epithelial cells [110–112]. In addition, mice lacking MyD88 are more susceptible to S. aureus infection suggesting that MyD88-dependent TLR2 signaling results in productive induction of immune responses. However, deletion of TLR2 did not completely abrogate induction of cytokines in macrophages, while deletion of TLR4 had no effect. These results suggest that additional PRRs, other than TLR2 and TLR4, detect S. aureus. Recent evidence points to a role for NLRs in recognizing S. aureus in macrophages due to the action of pore-forming toxins [113]. Owing to this efficient recognition of bacterial PAMPs, virtually all humans carry antibodies that recognize and opsonize S. aureus, and normal human serum promotes

Journal of Pathogens uptake by neutrophils. However, this does not always lead to bacterial killing. Instead, neutrophils and other phagocytic cells can be destroyed by the invasive strains [114, 115]. TLR2 activation leads to a proinflammatory response that recruits neutrophils, monocytes, T cells, and B cells to the infection site. In many cases, this response can be enough to clear the infection. Additionally, S. aureus is susceptible to surfactant, and opsonization of bacteria by SP-A can promote bacterial clearance [116]. S. aureus encodes a number of adhesins that play important roles in the pathogenesis of pneumonia. Invasion of epithelial cells involves fibronectin-binding protein and adhesins that have been specifically linked with invasive strains of Staphylococcus [117]. Invasion of these cells may be at least in part responsible for the ability of the bacteria to persistently colonize, as intracellular bacteria often evade host immunity [118]. Following invasion of human type II epithelial cells, there is little cell death in vitro [119]. In the intracellular environment, Staphylococci upregulate genes involved in iron scavenging and virulence, including several exotoxins, while simultaneously downregulating surface expressed protein A and the virulence associated transcription factor AgrA. Invasion of S. aureus into nonphagocytic cells was initially demonstrated in cultured mammary epithelial cells [120, 121]. These experiments established that intracellular bacteria could escape the endosome and induce apoptosis of epithelial cells. More recently, S. aureus was shown to survive inside activated neutrophils and induce pyroptosis following escape from the phagolysosome [115]. Invasion of epithelial cells can be accomplished through interactions between bacterial fibronectin binding protein and β1-integrins which promote Src protein-tyrosine kinase signaling to internalize the bacterium [122–124]. In addition, Staphylococcus adherence to epithelial cells is enhanced following viral infection which causes increased expression of host cell receptors such as ICAM-1 and downregulation of TLR2. This likely allows increased invasion and reduced NF-κB activation by the airway epithelial cells. S. aureus invasion of type II cells activates IFN-I. This activation is caused by the recognition of the conserved, multifunctional, secreted virulence factor protein A through its repeated IgG-binding domains [125]. Activation of IFNI leads to IL-6 and TNF-α production but instead of benefitting the host, IFN-I is detrimental during S. aureus infection. Mice lacking the IFN-I receptor, IFNAR, are more resistant to Staphylococcal pneumonia, a phenotype that correlates with a reduction in neutrophils and an increase in dendritic cells recruited to the lungs. This effect was found to be independent of the adaptor TRIF and IFN-β, while dependent on STAT-3. Thus, even though it is highly stimulatory through PRRs, Staphylococcal protein A leads to evasion of host immunity. Protein A is a major virulence factor, secreted by all pathogenic strains and required for the development of pneumonia in murine models [126]. Protein A binds host immunoglobulin with high affinity, thereby preventing its activation of Fc-receptor signaling. Protein A has long been appreciated for its role in virulence due to this activity, as

7 well as interfering with the opsonization of Staphylococci, blocking phagocytosis, and disabling complement fixation by the classical pathway. In addition, protein A binds with high affinity to TNF-R1, a receptor for the proinflammatory cytokine TNF-α, on bronchial epithelial cells which leads to the recruitment of neutrophils to the lungs [127]. All of these interactions are mediated through the repeated IgG binding domains [128]. Thus the bacteria use a single protein to both evade immunity and establish a replicative niche in the lungs from inside and outside of host cells. Extracellular bacteria produce many pore-forming toxins that specifically target leukocytes, some of which are unique to more virulent strains of S. aureus, while others are common to all of them [129–131]. Hemolysin is a wellconserved toxin that plays important roles during respiratory S. aureus infection. Hemolysin insertion into the plasma membrane of target cells creates a pore that alters ion gradients and membrane integrity, triggering cell death. Upon forming a pore in the smooth muscle cells that drive peristalsis, calcium release reduces contraction, allowing S. aureus access to the epithelial layer. There, hemolysin induces release of calcium from type II cells, leading to upregulation of IL-6 and prostaglandin production and, subsequently, inflammation [132, 133]. High affinity binding of α-hemolysin to type II epithelial cells is achieved through protein-protein interactions with ADAM-10, which not only leads to cell lysis, but also initiates signaling events that result in disruption of focal adhesions in the epithelial layer [134]. Once the bacterium accesses the alveolar spaces, S. aureus establishes a replicative niche, rapidly forming bacterial colonies that appear resistant to host neutrophils and other inflammatory cells [126]. Thus the activity of hemolysin paradoxically triggers inflammation while promoting invasion. Other exotoxins are epidemiologically linked to invasive CA-MRSA strains. For example, β-toxin of S. aureus binds syndecan-1, is internalized, then renders the host cell more vulnerable to other pathogen encoded toxins [135]. In addition, the toxin increases vascular permeability and edema in the lung which exacerbates lung injury and inflammation. Although not all pathogenic S. aureus strains carry βtoxin, there appears to be a correlation between enhanced capacity for respiratory infections and the presence of the β-toxin gene [108]. CA-MRSA strains express an additional virulence factor termed Panton-Valentine Leukocidin (PVL), a toxin that assembles into pore-forming octamers on the surface of host cells. Association between PVL expression and the pathogenesis of Staphylococcal pneumonia appears evident in the human population, but a definitive role in virulence in mouse and rabbit models of infection is controversial. When PVL is overexpressed, it appears to promote pneumonia in a mouse model [136]. However, when expressed at endogenous levels, PVL is not required for invasive Staphylococcal disease [137]. Nevertheless, PVL possesses potent membrane lysing activity on human neutrophils [138, 139]. In addition to this well-characterized activity, PVL also modulates signaling through TLR2 [140]. Purified PVL toxin is sufficient to cause TLR2- and CD14dependent inflammatory responses in the lungs following

8 intranasal inoculation, suggesting multiple roles for TLR2 in responding to S. aureus invasion of the lung [137]. 4.3. Yersinia pestis. Similar to F. tularensis, Yersinia pestis is a Gram-negative coccobacillus and is naturally transmitted to mammalian hosts by an arthropod vector [141]. Transmission via fleabite results in bubonic plague that can spread from the lymph to the blood, where organisms can then reach the lungs and cause secondary pneumonic plague. Once in the lung, organisms can be spread from person to person via aerosol droplets resulting in primary pneumonic plague, an acute bronchopneumonia. Pneumonic plague presents as a biphasic disease in that during the first 24– 36 hours of infection little inflammation is observed. The lung environment then abruptly turns proinflammatory, accompanied by rapid bacterial growth and tissue necrosis [142]. Pulmonary infection ultimately results in a patchy bronchopneumonia containing necrotic lesions composed of fibrin, neutrophils, and bacterial colonies [143]. In humans, symptoms include fever, headache, weakness, bloody sputum, and dyspnea. If left untreated, the infection is nearly always fatal. Y. pestis has acquired mechanisms to modify its LPS structure in response to temperature which prevents recognition of the bacterium by TLR4. When grown at lower temperatures (21–27◦ C), Y. pestis expresses a mixture of tri-acyl, tetra-acyl, penta-acyl, and hexa-acylated lipid A structures which may be beneficial for growth in this environment [144, 145]. However, when grown at the mammalian body temperature (37◦ C), tri-acyl and tetraacyl lipid A structures predominate, with no detectable hexa-acylation. Consequently, LPS isolated from bacteria grown at 37◦ C does not stimulate TLR4, and NF-κB is not activated in human inflammatory cells, thereby delaying production of TNF-α and IL-8 [146–148]. LPS isolated from bacteria grown at 37◦ C also inhibits TLR4 activation. Shown in mixing experiments, Y. pestis LPS from bacteria grown at 37◦ C can suppress TLR4 activation elicited from normally proinflammatory LPS [149]. Similar results are seen in dendritic cells, where it has also been shown that tetra-acylated LPS inhibits cell signaling through TLR2 and TLR9 and inhibits upregulation of the Costimulatory molecules MHC-II, CD40, and CD86 [150]. Together, the data demonstrate that LPS modulates TLR signaling through multiple mechanisms and is of central importance to Y. pestis virulence. Yersinia pestis also uses a type III secretion system (T3SS) to control inflammatory responses during infection through the injection of Yersinia outer proteins (Yops) into the host cell cytosol. Injection of Yops blocks phagocytic uptake by neutrophils, macrophages, and dendritic cells and ultimately causes host cell death [151–153]. This effect has historically been attributed to interactions with macrophages and neutrophils, however, the T3SS is active against epithelial cells and lymphocytes in vitro, suggesting that it is positioned to play multiple roles in vivo. Mutants that lack the T3SS are avirulent in pneumonic plague models, where they fail to evade early innate immune responses in the lung [154, 155].

Journal of Pathogens Depletion of TNF-α but not IL-1β causes an increase in sensitivity of mice to Y. pestis lacking YopH, but not wild type bacteria, suggesting that YopH may impact the ability of the host to induce NF-κB responses. Another type III effector protein, YopJ, has long been known to affect NFκB responses in macrophages. Recently, YopJ was shown to have similar activity when injected into bronchial epithelial cells where it reduced NF-κB regulated gene expression, suggesting that this virulence factor may help prevent unwanted inflammatory responses during the early stages of infection [156]. However, YopJ is relatively dispensable for virulence during pneumonic plague, suggesting that additional mechanisms for suppressing NF-κB regulated genes in alveolar macrophages and epithelial cells dominate during infection [157]. Effective T3S into phagocytic and epithelial cells has been shown to be dependent on the adhesive properties conferred by membrane proteins. Three proteins have been identified in Y. pestis that contribute this activity: Ail, Pla, and Psa. Ail (attachment-invasion locus) mediates binding to fibronectin, a component of the extracellular matrix [158]. Pla (plasminogen activator) has proteolytic and adhesive properties that also mediate binding to the extracellular matrix and perhaps other receptors on alveolar macrophages and dendritic cells [159–161]. Psa (pH 6 antigen) fimbriae bind to phosphatidylcholine, a component of cell membranes and surfactant, and to β1-linked, galactosyl-linked residues in glycosphingolipids [162, 163]. Psa appears to be more important for binding alveolar epithelial cells than to macrophages indicating that it may play a central role in penetration of the airway epithelium [164]. However, Psa is relatively dispensable for virulence during pneumonic plague, whereas Pla and Ail are essential [164]. Loss of all of these factors markedly reduces Yop-induced cytotoxicity towards target cells and attenuates virulence [165–168]. Y. pestis has a rough LPS structure and does not synthesize an O-antigen domain. Instead, bacteria utilize the multifunctional proteins mentioned above to resist killing by host antimicrobial molecules. Ail is highly expressed on the bacterial membrane at 26◦ C and 37◦ C and confers resistance to complement-mediated killing by serum derived from humans, rats, rabbits, sheep, goats, and guinea pigs but is dispensable for resistance to mouse serum [169]. Accordingly, strains lacking ail are highly attenuated in a rat model of pneumonic plague while in mice, ail mutants result in an increase in mean time to death, perhaps indicating a role for adhesion and internalization in vivo [170]. In addition, antimicrobial peptides such as cathelicidin and β-defensin have antimicrobial activity against attenuated strains of Y. pestis in vitro, and expression of the surface located virulence factors Pla and CaF1 (Capsular protein F1) influence susceptibility to these peptides [171]. Pla is a serine protease with broad spectrum activity that plays an essential role in the development of pneumonic plague. Pla-catalyzed cleavage of cationic antimicrobial peptides provides a mechanism whereby Y. pestis can cleave and inactivate CAMPs [171]. Paradoxically, expression of CaF1, which forms antiphagocytic pili on the Y. pestis cell surface at 37◦ C, can reduce the protective effects elicited by Pla,

Journal of Pathogens

9

Francisella tularensis

Staphylococcus aureus IL-8 MCP-1 MIP1-α

hBD1 hBD2 MAC

Immunoglobulin

Yersinia pestis

IFN-α/β IL-6 Apoptosis TNF-α

CAMPs

MAC

IFN-1 IL-1β

NF-κB hBD3

NF-κB Yops

TLR4 TNF-R1

Lysis

IKK-β

Ca2+

Sepsis

MAC

Sepsis

IL-1β IL-18 Pyroptosis IFN-γR

STAT1 pro-IL-1β L

NADPH oxidase

Immunoglobulin Sepsis

MAC

TLR2

Apoptosis

Lysis Pyroptosis

Caspase-1

TLR4

Caspase-3

TLR4

Lysis

Yops Yops

IL-1β

NF-κB

- Mucous layer

- Type II pneumocyte

- Blood

- Macrophage

- Bacteria

IκB-Ub

ROS PICD

TLR2

ROS L

NF-κB

- Neutrophil

- Endothelial cell - Type I pneumocyte and/or bronchial epithelial cell

IκB-Ub

TLR4

TLR4

- Phagosome L

- Lysosome

Figure 1: Disruption of airway defenses by Francisella, Staphylococcus, and Yersinia. Summary of host pathogen interactions used by these bacteria to modulate innate immune responses and invade the airway.

likely through steric interference or alteration of substrate specificity. Pla may also directly mediate serum resistance due to its proteolytic activity on C3 [160]. Yersinia species are thought to be capable of invading epithelial cells through interactions between one or more adhesins and host cell β1 integrins [172]. At least three pathways of invasion have been suggested based on interactions between enteropathogenic Yersiniae and epithelial cells, the most efficient of which, mediated by the protein Invasin, is not likely not to occur in Y. pestis because this gene is not expressed [173]. Nevertheless, invasion of the bronchial or type II alveolar epithelial cells is an attractive model by which Y. pestis would successfully penetrate the epithelial barrier without causing inflammation at early stages of infection. Alternative mechanisms for how bacteria invade the epithelium involve the action of one or more toxins

produced by extracellular bacteria. In support of this model, Pla is required for Y. pestis to invade the lung parenchyma, suggesting that it may have a role in enhancing penetration of the alveolar epithelium [168]. Even under conditions that support high level, simultaneous expression of virulence factors that suppress phagocytosis and contribute to the extracellular lifecycle of Y. pestis, a small percentage of bacteria are engulfed by phagocytes in vitro [174]. Intracellular bacteria are equipped to resist antimicrobial activity and proliferate even in IFN-γ activated macrophages [175, 176]. Once phagocytosed by macrophages, bacteria prevent the acidification of vacuoles and begin replicating independent of the T3SS [177, 178]. Replication in activated macrophages requires the protein RipA which directly reduces NO levels without modulating iNOS expression [175]. Intracellular survival is also

10 dependent on phoPQ (a two-component signal transduction system that responds to low [Mg+2 ]), ugd, pmrK (predicted phagosomal antimicrobial peptide resistance genes), and mgtC (a low-Mg2+ induced gene) which are important for the early intracellular survival of Y. pestis [179–181]. In addition, antibody opsonization of Y. pestis promotes phagocytic uptake, but the bacteria are not killed by macrophages and bacterial clearance by opsonizing antibodies requires neutrophils [182–184]. Together, the data suggest that survival in alveolar macrophages and perhaps also epithelial cells lining the airway may be an important virulence mechanism for invasive strategies utilized during pneumonic plague [164]. In addition to inhibiting bacteria from being internalized, T3S by extracellular bacteria also inhibits ROS production in phagocytic cells which is required to eliminate intracellular bacteria [153]. Neutrophils are resistant to Yopinduced apoptosis and inhibition of ROS production appears to prevent cells from undergoing phagocytosis-induced cell death (PICD), a mechanism used by neutrophils to contain infection and resolve inflammation [185, 186]. Thus, the data support a model whereby T3S blocking antibodies prevent Yop injection into neutrophils, allowing ROS production and subsequent killing of intra- and extracellular bacteria.

5. Conclusions The mammalian lower respiratory tract is largely protected by the functions of airway epithelial cells. These cells are sentinels, orchestrating recruitment, activation and deactivation of inflammatory cells when microbes attempt to invade the lung. Through the continuous production of mucin and surfactant loaded with antimicrobial molecules, potentially harmful bacteria are trapped and cleared. If bacteria can avoid or resist these normally protective mechanisms, they need only to destroy or cross these cells to establish a replicative niche before an onslaught of inflammatory cells arrives. Yersinia, Francisella, and Staphylococcus, three bacterial pathogens with the capability to cause lower respiratory tract infection and acute pneumonia, possess multiple mechanisms for penetrating the epithelium and evading innate immunity, many of which exploit these defense mechanisms to promote virulence (Figure 1). In common between these and other bacterial pneumonias is the use of cell surface structures that evade recognition and resist the antimicrobial defenses of the airway epithelium. Bacterial pathogens have enormous capacity for continuous and rapid evolution allowing organisms to adapt in order to further tip the balance of host-pathogen interactions in favor of invasion across the epithelium, replication, and disease.

Acknowledgments The authors regret that due to space limitations they were unable to discuss all of the research on Francisella, Staphylococcus, and Yersinia, some of which represent findings critical to pathogenesis in humans. This paper is dedicated to the memory of Daniel P. Saathoff. The authors are grateful to

Journal of Pathogens David Bland for critical comments on the paper. N. A. Eisele is supported by NIH/NIGMS Cellular and Molecular Biology training grant T32 GM008396.

References [1] M. C. Rose, T. J. Nickola, and J. A. Voynow, “Airway mucus obstruction: mucin glycoproteins, MUC gene regulation and goblet cell hyperplasia,” American Journal of Respiratory Cell and Molecular Biology, vol. 25, no. 5, pp. 533–537, 2001. [2] J. A. Davies and D. R. Garrod, “Molecular aspects of the epithelial phenotype,” BioEssays, vol. 19, no. 8, pp. 699–704, 1997. [3] M. R. Knowles and R. C. Boucher, “Mucus clearance as a primary innate defense mechanism for mammalian airways,” Journal of Clinical Investigation, vol. 109, no. 5, pp. 571–577, 2002. [4] L. Yang, J. Johansson, R. Ridsdale et al., “Surfactant protein B propeptide contains a saposin-like protein domain with antimicrobial activity at low pH,” Journal of Immunology, vol. 184, no. 2, pp. 975–983, 2010. [5] H. Fehrenbach, “Alveolar epithelial type II cell: defender of the alveolus revisited,” Respiratory Research, vol. 2, no. 1, pp. 33–46, 2001. [6] R. J. Mason, “Biology of alveolar type II cells,” Respirology, vol. 11, no. 1, pp. S12–S15, 2006. [7] G. Guadiz, L. A. Sporn, R. A. Goss, S. O. Lawrence, V. J. Marder, and P. J. Simpson-Haidaris, “Polarized secretion of fibrinogen by lung epithelial cells,” American Journal of Respiratory Cell and Molecular Biology, vol. 17, no. 1, pp. 60– 69, 1997. [8] A. J. Ratner, K. R. Hippe, J. L. Aguilar, M. H. Bender, A. L. Nelson, and J. N. Weiser, “Epithelial cells are sensitive detectors of bacterial pore-forming toxins,” Journal of Biological Chemistry, vol. 281, no. 18, pp. 12994–12998, 2006. [9] A. Punturieri, R. S. Alviani, T. Polak, P. Copper, J. Sonstein, and J. L. Curtis, “Specific engagement of TLR4 or TLR3 does not lead to IFN-β-mediated innate signal amplification and STAT1 phosphorylation in resident murine alveolar macrophages,” Journal of Immunology, vol. 173, no. 2, pp. 1033–1042, 2004. [10] L. J. Quinton, M. R. Jones, B. E. Robson, B. T. Simms, J. A. Whitsett, and J. P. Mizgerd, “Alveolar epithelial STAT3, IL-6 family cytokines, and host defense during Escherichia coli pneumonia,” American Journal of Respiratory Cell and Molecular Biology, vol. 38, no. 6, pp. 699–706, 2008. [11] S. Rosseau, J. Selhorst, K. Wiechmann et al., “Monocyte migration through the alveolar epithelial barrier: adhesion molecule mechanisms and impact of chemokines,” Journal of Immunology, vol. 164, no. 1, pp. 427–435, 2000. [12] T. R. Martin, N. Hagimoto, M. Nakamura, and G. MatuteBello, “Apoptosis and epithelial injury in the lungs,” Proceedings of the American Thoracic Society, vol. 2, no. 3, pp. 214– 220, 2005. [13] M. Vareille, E. Kieninger, M. R. Edwards, and N. Regamey, “The airway epithelium: soldier in the fight against respiratory viruses,” Clinical Microbiology Reviews, vol. 24, no. 1, pp. 210–229, 2011. [14] T. Balamayooran, G. Balamayooran, and S. Jeyaseelan, “Toll-like receptors and NOD-like receptors in pulmonary antibacterial immunity,” Innate Immunity, vol. 16, no. 3, pp. 201–210, 2010.

Journal of Pathogens [15] X. Zhao, S. Nozell, Z. Ma, and E. N. Benveniste, “The interferon-stimulated gene factor 3 complex mediates the inhibitory effect of interferon-β on matrix metalloproteinase-9 expression,” FEBS Journal, vol. 274, no. 24, pp. 6456– 6468, 2007. [16] S. I. Miller, R. K. Ernst, and M. W. Bader, “LPS, TLR4 and infectious disease diversity,” Nature Reviews Microbiology, vol. 3, no. 1, pp. 36–46, 2005. [17] Z. Zhou, O. J. Hamming, N. Ank, S. R. Paludan, A. L. Nielsen, and R. Hartmann, “Type III interferon (IFN) induces a type I IFN-like response in a restricted subset of cells through signaling pathways involving both the Jak-STAT pathway and the mitogen-activated protein kinases,” Journal of Virology, vol. 81, no. 14, pp. 7749–7758, 2007. [18] N. Ank and S. R. Paludan, “Type III IFNs: new layers of complexity in innate antiviral immunity,” BioFactors, vol. 35, no. 1, pp. 82–87, 2009. [19] J. Fan, R. S. Frey, and A. B. Malik, “TLR4 signaling induces TLR2 expression in endothelial cells via neutrophil NADPH oxidase,” Journal of Clinical Investigation, vol. 112, no. 8, pp. 1234–1243, 2003. [20] G. Soong, B. Reddy, S. Sokol, R. Adamo, and A. Prince, “TLR2 is mobilized into an apical lipid raft receptor complex to signal infection in airway epithelial cells,” Journal of Clinical Investigation, vol. 113, no. 10, pp. 1482–1489, 2004. [21] S. Bunk, S. Sigel, D. Metzdorf et al., “Internalization and coreceptor expression are critical for TLR2-mediated recognition of lipoteichoic acid in human peripheral blood,” Journal of Immunology, vol. 185, no. 6, pp. 3708–3717, 2010. [22] M. Choi, Z. Wang, T. Ban et al., “A selective contribution of the RIG-I-like receptor pathway to type I interferon responses activated by cytosolic DNA,” Proceedings of the National Academy of Sciences of the United States of America, vol. 106, no. 42, pp. 17870–17875, 2009. [23] A. Takaoka, Z. Wang, M. K. Choi et al., “DAI (DLM-1/ZBP1) is a cytosolic DNA sensor and an activator of innate immune response,” Nature, vol. 448, no. 7152, pp. 501–505, 2007. [24] K. J. Ishii, T. Kawagoe, S. Koyama et al., “TANK-binding kinase-1 delineates innate and adaptive immune responses to DNA vaccines,” Nature, vol. 451, no. 7179, pp. 725–729, 2008. [25] Z. Wang, M. K. Choi, T. Ban et al., “Regulation of innate immune responses by DAI (DLM-1/ZBP1) and other DNAsensing molecules,” Proceedings of the National Academy of Sciences of the United States of America, vol. 105, no. 14, pp. 5477–5482, 2008. [26] T. Melkamu, D. Squillace, H. Kita, and S. M. O’Grady, “Regulation of TLR2 expression and function in human airway epithelial cells,” Journal of Membrane Biology, vol. 229, no. 2, pp. 101–113, 2009. [27] D. M. Morens, J. K. Taubenberger, and A. S. Fauci, “Predominant role of bacterial pneumonia as a cause of death in pandemic influenza: implications for pandemic influenza preparedness,” Journal of Infectious Diseases, vol. 198, no. 7, pp. 962–970, 2008. [28] S. Herold, M. Steinmueller, W. Von Wulffen et al., “Lung epithelial apoptosis in influenza virus pneumonia: the role of macrophage-expressed TNF-related apoptosis-inducing ligand,” Journal of Experimental Medicine, vol. 205, no. 13, pp. 3065–3077, 2008. [29] C. L. Small, C. R. Shaler, S. McCormick et al., “Influenza infection leads to increased susceptibility to subsequent bacterial superinfection by impairing NK cell responses in

11

[30]

[31]

[32]

[33]

[34]

[35]

[36]

[37]

[38]

[39]

[40] [41]

[42]

[43]

[44]

[45]

the lung,” Journal of Immunology, vol. 184, no. 4, pp. 2048– 2056, 2010. E. DiMango, H. J. Zar, R. Bryan, and A. Prince, “Diverse Pseudomonas aeruginosa gene products stimulate respiratory epithelial cells to produce interleukin-8,” Journal of Clinical Investigation, vol. 96, no. 5, pp. 2204–2210, 1995. A. Marfaing-Koka, O. Devergne, G. Gorgone et al., “Regulation of the production of the RANTES chemokine by endothelial cells: synergistic induction by IFN-γ plus TNFα and inhibition by IL-4 and IL-13,” Journal of Immunology, vol. 154, no. 4, pp. 1870–1878, 1995. C. Stellato, L. A. Beck, G. A. Gorgone et al., “Expression of the chemokine RANTES by a human bronchial epithelial cell line: modulation by cytokines and glucocorticoids,” Journal of Immunology, vol. 155, no. 1, pp. 410–418, 1995. Y. Lin, M. Zhang, and P. F. Barnes, “Chemokine production by a human alveolar epithelial cell line in response to Mycobacterium tuberculosis,” Infection and Immunity, vol. 66, no. 3, pp. 1121–1126, 1998. A. Belaaouaj, “Neutrophil elastase-mediated killing of bacteria: lessons from targeted mutagenesis,” Microbes and Infection, vol. 4, no. 12, pp. 1259–1264, 2002. S. Griffin, C. C. Taggart, C. M. Greene, S. O’Neill, and N. G. McElvaney, “Neutrophil elastase up-regulates human βdefensin-2 expression in human bronchial epithelial cells,” FEBS Letters, vol. 546, no. 2-3, pp. 233–236, 2003. K. K. Adkins, T. D. Levan, R. L. Miesfeld, and J. W. Bloom, “Glucocorticoid regulation of GM-CSF: evidence for transcriptional mechanisms in airway epithelial cells,” American Journal of Physiology, vol. 275, no. 2, pp. L372– L378, 1998. G. K. Paterson and C. J. Orihuela, “Pneumococci: immunology of the innate host response,” Respirology, vol. 15, no. 7, pp. 1057–1063, 2010. R. Podschun and U. Ullmann, “Klebsiella spp. as nosocomial pathogens: epidemiology, taxonomy, typing methods, and pathogenicity factors,” Clinical Microbiology Reviews, vol. 11, no. 4, pp. 589–603, 1998. S. W. Kim, C. H. Choi, D. C. Moon et al., “Serum resistance of Acinetobacter baumannii through the binding of factor H to outer membrane proteins,” FEMS Microbiology Letters, vol. 301, no. 2, pp. 224–231, 2009. J. E. Foley and N. C. Nieto, “Tularemia,” Veterinary Microbiology, vol. 140, no. 3-4, pp. 332–338, 2010. M. K. McLendon, M. A. Apicella, and L. A. H. Allen, “Francisella tularensis: taxonomy, genetics, and immunopathogenesis of a potential agent of biowarfare,” Annual Review of Microbiology, vol. 60, pp. 167–185, 2006. L. D. Thomas and W. Schaffner, “Tularemia pneumonia,” Infectious Disease Clinics of North America, vol. 24, no. 1, pp. 43–55, 2010. A. Balagopal, A. S. MacFarlane, N. Mohapatra, S. Soni, J. S. Gunn, and L. S. Schlesinger, “Characterization of the receptor-ligand pathways important for entry and survival of Francisella tularensis in human macrophages,” Infection and Immunity, vol. 74, no. 9, pp. 5114–5125, 2006. D. L. Clemens, B. Y. Lee, and M. A. Horwitz, “Francisella tularensis enters macrophages via a novel process involving pseudopod loops,” Infection and Immunity, vol. 73, no. 9, pp. 5892–5902, 2005. H. Geier and J. Celli, “Phagocytic receptors dictate phagosomal escape and intracellular proliferation of Francisella tularensis,” Infection and Immunity, vol. 79, no. 6, pp. 2204– 2214, 2011.

12 [46] L. S. D. Anthony, R. D. Burke, and F. E. Nano, “Growth of Francisella spp. in rodent macrophages,” Infection and Immunity, vol. 59, no. 9, pp. 3291–3296, 1991. [47] I. Golovliov, V. Baranov, Z. Krocova, H. Kovarova, and A. Sj¨ostedt, “An attenuated strain of the facultative intracellular bacterium Francisella tularensis can escape the phagosome of monocytic cells,” Infection and Immunity, vol. 71, no. 10, pp. 5940–5950, 2003. [48] C. Checroun, T. D. Wehrly, E. R. Fischer, S. F. Hayes, and J. Celli, “Autophagy-mediated reentry of Francisella tularensis into the endocytic compartment after cytoplasmic replication,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 39, pp. 14578– 14583, 2006. [49] J. R. Barker, A. Chong, T. D. Wehrly et al., “The Francisella tularensis pathogenicity island encodes a secretion system that is required for phagosome escape and virulence,” Molecular Microbiology, vol. 74, no. 6, pp. 1459–1470, 2009. [50] F. E. Nano, N. Zhang, S. C. Cowley et al., “A Francisella tularensis pathogenicity island required for intramacrophage growth,” Journal of Bacteriology, vol. 186, no. 19, pp. 6430– 6436, 2004. [51] C. M. Lauriano, J. R. Barker, S. S. Yoon et al., “MgIA regulates transcription of virulence factors necessary for Francisella tularensis intraamoebae and intramacrophage survival,” Proceedings of the National Academy of Sciences of the United States of America, vol. 101, no. 12, pp. 4246–4249, 2004. [52] R. D. Pechous, T. R. McCarthy, and T. C. Zahrt, “Working toward the future: insights into Francisella tularensis pathogenesis and vaccine development,” Microbiology and Molecular Biology Reviews, vol. 73, no. 4, pp. 684–711, 2009. [53] K. L. Elkins, T. R. Rhinehart-Jones, S. J. Culkin, D. Yee, and R. K. Winegar, “Minimal requirements for murine resistance to infection with Francisella tularensis LVS,” Infection and Immunity, vol. 64, no. 8, pp. 3288–3293, 1996. [54] D. A. Leiby, A. H. Fortier, R. M. Crawford, R. D. Schreiber, and C. A. Nacy, “In vivo modulation of the murine immune response to Francisella tularensis LVS by administration of anticytokine antibodies,” Infection and Immunity, vol. 60, no. 1, pp. 84–89, 1992. [55] J. E. Darnell, I. M. Kerr, and G. R. Stark, “Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins,” Science, vol. 264, no. 5164, pp. 1415–1421, 1994. [56] T. Polsinelli, M. S. Meltzer, and A. H. Fortier, “Nitric oxide-independent killing of Francisella tularensis by IFNγ- stimulated murine alveolar macrophages,” Journal of Immunology, vol. 153, no. 3, pp. 1238–1245, 1994. [57] H. Lindgren, I. Golovliov, V. Baranov, R. K. Ernst, M. Telepnev, and A. Sj¨ostedt, “Factors affecting the escape of Francisella tularensis from the phagolysosome,” Journal of Medical Microbiology, vol. 53, no. 10, pp. 953–958, 2004. [58] M. Santic, M. Molmeret, and Y. Abu Kwaik, “Modulation of biogenesis of the Francisella tularensis subsp. novicidacontaining phagosome in quiescent human macrophages and its maturation into a phagolysosome upon activation by IFNγ,” Cellular Microbiology, vol. 7, no. 7, pp. 957–967, 2005. [59] J. A. Edwards, D. Rockx-Brouwer, V. Nair, and J. Celli, “Restricted cytosolic growth of Francisella tularensis subsp. tularensis by IFN-γ activation of macrophages,” Microbiology, vol. 156, no. 2, pp. 327–339, 2010. [60] K. C. Nallaparaju, J. -J. Yu, S. A. Rodriguez et al., “Evasion of IFN-γ signaling by Francisella novicida is dependent upon

Journal of Pathogens

[61]

[62]

[63]

[64]

[65]

[66]

[67]

[68]

[69]

[70]

[71]

[72]

[73]

[74]

Francisella outer membrane protein C,” PLoS ONE, vol. 6, no. 3, Article ID e18201, 2011. J. S. Gunn and R. K. Ernst, “The structure and function of Francisella lipopolysaccharide,” Annals of the New York Academy of Sciences, vol. 1105, pp. 202–218, 2007. A. L. Abplanalp, I. R. Morris, B. K. Parida, J. M. Teale, and M. T. Berton, “TLR-dependent control of Francisella tularensis infection and host inflammatory responses,” PLoS ONE, vol. 4, no. 11, Article ID e7920, 2009. J. H. Barker, J. Weiss, M. A. Apicella, and W. M. Nauseef, “Basis for the failure of Francisella tularensis lipopolysaccharide to prime human polymorphonuclear leukocytes,” Infection and Immunity, vol. 74, no. 6, pp. 3277–3284, 2006. ` W. Chen, R. KuoLee, H. Shen, M. Busa, and J. W. Conlan, “Toll-like receptor 4 (TLR4) does not confer a resistance advantage on mice against low-dose aerosol infection with virulent type A Francisella tularensis,” Microbial Pathogenesis, vol. 37, no. 4, pp. 185–191, 2004. A. M. Hajjar, M. D. Harvey, S. A. Shaffer et al., “Lack of in vitro and in vivo recognition of Francisella tularensis subspecies lipopolysaccharide by Toll-like receptors,” Infection and Immunity, vol. 74, no. 12, pp. 6730–6738, 2006. E. Vinogradov, M. B. Perry, and J. W. Conlan, “Structural analysis of Francisella tularensis lipopolysaccharide,” European Journal of Biochemistry, vol. 269, no. 24, pp. 6112–6118, 2002. A. I. Due˜nas, M. Aceves, A. Ordu˜na, R. D´ıaz, M. S´anchez Crespo, and C. Garc´ıa-Rodr´ıguez, “Francisella tularensis LPS induces the production of cytokines in human monocytes and signals via Toll-like receptor 4 with much lower potency than E. coli LPS,” International Immunology, vol. 18, no. 5, pp. 785–795, 2006. N. J. Phillips, B. Schilling, M. K. McLendon, M. A. Apicella, and B. W. Gibson, “Novel modification of lipid A of Francisella tularensis,” Infection and Immunity, vol. 72, no. 9, pp. 5340–5348, 2004. X. Wang, S. C. McGrath, R. J. Cotter, and C. R. H. Raetz, “Expression cloning and periplasmic orientation of the Francisella novicida lipid A 4 -phosphatase LpxF,” Journal of Biological Chemistry, vol. 281, no. 14, pp. 9321–9330, 2006. X. Wang, A. A. Ribeiro, Z. Guan, S. N. Abraham, and C. R. H. Raetz, “Attenuated virulence of a Francisella mutant lacking the lipid a 4 -phosphatase,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 10, pp. 4136–4141, 2007. A. Lembo, M. Pelletier, R. Iyer et al., “Administration of a synthetic TLR4 agonist protects mice from pneumonic tularemia,” Journal of Immunology, vol. 180, no. 11, pp. 7574– 7581, 2008. D. Kanistanon, A. M. Hajjar, M. R. Pelletier et al., “A Francisella mutant in lipid A carbohydrate modification elicits protective immunity,” PLoS Pathogens, vol. 4, no. 2, article e24, 2008. X. H. Lai, R. L. Shirley, L. Crosa et al., “Mutations of Francisella novicida that alter the mechanism of its phagocytosis by murine macrophages,” PLoS ONE, vol. 5, no. 7, Article ID e11857, 2010. C. D. Clay, S. Soni, J. S. Gunn, and L. S. Schlesinger, “Evasion of complement-mediated lysis and complement C3 deposition are regulated by Francisella tularensis lipopolysaccharide O antigen,” Journal of Immunology, vol. 181, no. 8, pp. 5568– 5578, 2008.

Journal of Pathogens [75] P. Gros, F. J. Milder, and B. J. C. Janssen, “Complement driven by conformational changes,” Nature Reviews Immunology, vol. 8, no. 1, pp. 48–58, 2008. [76] A. B. Nasr and G. R. Klimpe, “Subversion of complement activation at the bacterial surface promotes serum resistance and opsonophagocytosis of Francisella tularensis,” Journal of Leukocyte Biology, vol. 84, no. 1, pp. 77–85, 2008. [77] M. A. Apicella, D. M. B. Post, A. C. Fowler et al., “Identification, characterization and immunogenicity of an O-antigen capsular polysaccharide of Francisella tularensis,” PLoS ONE, vol. 5, no. 7, Article ID e11060, 2010. [78] S. R. Lindemann, K. Peng, M. E. Long et al., “Francisella tularensis Schu S4 O-antigen and capsule biosynthesis gene mutants induce early cell death in human macrophages,” Infection and Immunity, vol. 79, no. 2, pp. 581–594, 2011. [79] S. Sebastian, S. T. Dillon, J. G. Lynch et al., “A defined Oantigen polysaccharide mutant of Francisella tularensis live vaccine strain has attenuated virulence while retaining its protective capacity,” Infection and Immunity, vol. 75, no. 5, pp. 2591–2602, 2007. [80] J. Li, C. Ryder, M. Mandal et al., “Attenuation and protective efficacy of an O-antigen-deficient mutant of Francisella tularensis LVS,” Microbiology, vol. 153, no. 9, pp. 3141–3153, 2007. [81] S. Han, B. M. Bishop, and M. L. van Hoek, “Antimicrobial activity of human beta-defensins and induction by Francisella,” Biochemical and Biophysical Research Communications, vol. 371, no. 4, pp. 670–674, 2008. [82] M. Pazgier, D. M. Hoover, D. Yang, W. Lu, and J. Lubkowski, “Human β-defensins,” Cellular and Molecular Life Sciences, vol. 63, no. 11, pp. 1294–1313, 2006. [83] K. De Smet and R. Contreras, “Human antimicrobial peptides: defensins, cathelicidins and histatins,” Biotechnology Letters, vol. 27, no. 18, pp. 1337–1347, 2005. [84] P. K. Singh, H. P. Jia, K. Wiles et al., “Production of βdefensins by human airway epithelia,” Proceedings of the National Academy of Sciences of the United States of America, vol. 95, no. 25, pp. 14961–14966, 1998. [85] J. Harder, J. Bartels, E. Christophers, and J. M. Schr¨oder, “Isolation and characterization of human β-defensin-3, a novel human inducible peptide antibiotic,” Journal of Biological Chemistry, vol. 276, no. 8, pp. 5707–5713, 2001. [86] T. Henry and D. M. Monack, “Activation of the inflammasome upon Francisella tularensis infection: interplay of innate immune pathways and virulence factors,” Cellular Microbiology, vol. 9, no. 11, pp. 2543–2551, 2007. [87] M. Malik, C. S. Bakshi, B. Sahay, A. Shah, S. A. Lotz, and T. J. Sellati, “Toll-like receptor 2 is required for control of pulmonary infection with Francisella tularensis,” Infection and Immunity, vol. 74, no. 6, pp. 3657–3662, 2006. [88] T. Henry, A. Brotcke, D. S. Weiss, L. J. Thompson, and D. M. Monack, “Type I interferon signaling is required for activation of the inflammasome during Francisella infection,” Journal of Experimental Medicine, vol. 204, no. 5, pp. 987– 994, 2007. [89] D. S. Weiss, T. Henry, and D. M. Monack, “Francisella tularensis: activation of the inflammasome,” Annals of the New York Academy of Sciences, vol. 1105, pp. 219–237, 2007. [90] H. Li, S. Nookala, X. R. Bina, J. E. Bina, and F. Re, “Innate immune response to Francisella tularensis is mediated by TLR2 and caspase-1 activation,” Journal of Leukocyte Biology, vol. 80, no. 4, pp. 766–773, 2006. [91] L. E. Cole, A. Santiago, E. Barry et al., “Macrophage proinflammatory response to Francisella tularensis live vaccine

13

[92]

[93]

[94]

[95]

[96]

[97]

[98]

[99]

[100]

[101]

[102]

[103]

[104]

[105]

strain requires coordination of multiple signaling pathways,” Journal of Immunology, vol. 180, no. 10, pp. 6885–6891, 2008. L. E. Cole, K. A. Shirey, E. Barry et al., “Toll-like receptor 2mediated signaling requirements for Francisella tularensis live vaccine strain infection of murine macrophages,” Infection and Immunity, vol. 75, no. 8, pp. 4127–4137, 2007. S. Thakran, H. Li, C. L. Lavine et al., “Identification of Francisella tularensis lipoproteins that stimulate the toll-like receptor (TLR) 2/TLR1 heterodimer,” Journal of Biological Chemistry, vol. 283, no. 7, pp. 3751–3760, 2008. E. A. Medina, I. R. Morris, and M. T. Berton, “Phosphatidylinositol 3-kinase activation attenuates the TLR2-mediated macrophage proinflammatory cytokine response to Francisella tularensis live vaccine strain,” Journal of Immunology, vol. 185, no. 12, pp. 7562–7572, 2010. J. D. Hall, R. R. Craven, J. R. Fuller, R. J. Pickles, and T. H. Kawula, “Francisella tularensis replicates within alveolar type II epithelial cells in vitro and in vivo following inhalation,” Infection and Immunity, vol. 75, no. 2, pp. 1034–1039, 2007. S. Chakraborty, M. Monfett, T. M. Maier, J. L. Benach, D. W. Frank, and D. G. Thanassi, “Type IV pili in Francisella tularensis: roles of pilF and pilT in fiber assembly, host cell adherence, and virulence,” Infection and Immunity, vol. 76, no. 7, pp. 2852–2861, 2008. A. Melillo, D. D. Sledjeski, S. Lipski, R. M. Wooten, V. Basrur, and E. R. Lafontaine, “Identification of a Francisella tularensis LVS outer membrane protein that confers adherence to A549 human lung cells,” FEMS Microbiology Letters, vol. 263, no. 1, pp. 102–108, 2006. R. R. Craven, J. D. Hall, J. R. Fuller, S. Taft-Benz, and T. H. Kawula, “Francisella tularensis invasion of lung epithelial cells,” Infection and Immunity, vol. 76, no. 7, pp. 2833–2842, 2008. M. Gentry, J. Taormina, R. B. Pyles et al., “Role of primary human alveolar epithelial cells in host defense against Francisella tularensis infection,” Infection and Immunity, vol. 75, no. 8, pp. 3969–3978, 2007. M. Malik, C. S. Bakshi, K. McCabe et al., “Matrix metalloproteinase 9 activity enhances host susceptibility to pulmonary infection with type A and B strains of Francisella tularensis,” Journal of Immunology, vol. 178, no. 2, pp. 1013–1020, 2007. J. G. Moreland, J. S. Hook, G. Bailey, T. Ulland, and W. M. Nauseef, “Francisella tularensis directly interacts with the endothelium and recruits neutrophils with a blunted inflammatory phenotype,” American Journal of Physiology, vol. 296, no. 6, pp. L1076–L1084, 2009. R. KuoLee, G. Harris, J. W. Conlan, and W. Chen, “Role of neutrophils and NADPH phagocyte oxidase in host defense against respiratory infection with virulent Francisella tularensis in mice,” Microbes and Infection, vol. 13, no. 5, pp. 447–456, 2011. R. L. McCaffrey and L. A. H. Allen, “Pivotal advance: Francisella tularensis LVS evades killing by human neutrophils via inhibition of the respiratory burst and phagosome escape,” Journal of Leukocyte Biology, vol. 80, no. 6, pp. 1224–1230, 2006. J. H. Barker, R. L. McCaffrey, N. K. Baman, L. A. H. Allen, J. P. Weiss, and W. M. Nauseef, “The role of complement opsonization in interactions between F. tularensis subsp. novicida and human neutrophils,” Microbes and Infection, vol. 11, no. 8-9, pp. 762–769, 2009. T. J. Reilly, G. S. Baron, F. E. Nano, and M. S. Kuhlenschmidt, “Characterization and sequencing of a respiratory burstinhibiting acid phosphatase from Francisella tularensis,”

14

[106]

[107]

[108]

[109]

[110]

[111]

[112]

[113]

[114]

[115]

[116]

[117]

[118]

[119]

Journal of Pathogens Journal of Biological Chemistry, vol. 271, no. 18, pp. 10973– 10983, 1996. N. P. Mohapatra, S. Soni, M. V. S. Rajaram et al., “Francisella acid phosphatases inactivate the NADPH oxidase in human phagocytes,” Journal of Immunology, vol. 184, no. 9, pp. 5141– 5150, 2010. G. S. Schulert, R. L. McCaffrey, B. W. Buchan et al., “Francisella tularensis genes required for inhibition of the neutrophil respiratory burst and intramacrophage growth identified by random transposon mutagenesis of strain LVS,” Infection and Immunity, vol. 77, no. 4, pp. 1324–1336, 2009. B. A. Diep, H. A. Carleton, R. F. Chang, G. F. Sensabaugh, and F. Perdreau-Remington, “Roles of 34 virulence genes in the evolution of hospital- and community-associated strains of methicillin-resistant Staphylococcus aureus,” Journal of Infectious Diseases, vol. 193, no. 11, pp. 1495–1503, 2006. R. M. Klevens, M. A. Morrison, J. Nadle et al., “Invasive methicillin-resistant Staphylococcus aureus infections in the United States,” Journal of the American Medical Association, vol. 298, no. 15, pp. 1763–1771, 2007. O. Takeuchi, K. Hoshino, and S. Akira, “Cutting edge: TLR2deficient and MyD88-deficient mice are highly susceptible to Staphylococcus aureus infection,” Journal of Immunology, vol. 165, no. 10, pp. 5392–5396, 2000. J. E. Wang, P. F. Jørgensen, M. Alml¨of et al., “Peptidoglycan and lipoteichoic acid from Staphylococcus aureus induce tumor necrosis factor alpha, interleukin 6 (IL-6), and IL10 production in both T cells and monocytes in a human whole blood model,” Infection and Immunity, vol. 68, no. 7, pp. 3965–3970, 2000. I. S. Cheon, S. S. Woo, S. S. Kang et al., “Peptidoglycanmediated IL-8 expression in human alveolar type II epithelial cells requires lipid raft formation and MAPK activation,” Molecular Immunology, vol. 45, no. 6, pp. 1665–1673, 2008. P. Hruz, A. S. Zinkernagel, G. Jenikova et al., “NOD2 contributes to cutaneous defense against Staphylococcus aureus through α-toxin-dependent innate immune activation,” Proceedings of the National Academy of Sciences of the United States of America, vol. 106, no. 31, pp. 12873–12878, 2009. A. Shiratsuchi, K. Shimizu, I. Watanabe et al., “Auxiliary role for D-alanylated wall teichoic acid in Toll-like receptor 2mediated survival of Staphylococcus aureus in macrophages,” Immunology, vol. 129, no. 2, pp. 268–277, 2010. S. D. Kobayashi, K. R. Braughton, A. M. Palazzolo-Ballance et al., “Rapid neutrophil destruction following phagocytosis of Staphylococcus aureus,” Journal of Innate Immunity, vol. 2, no. 6, pp. 560–575, 2010. Z. Sever-Chroneos, A. Krupa, J. Davis et al., “Surfactant protein A (SP-A)-mediated clearance of Staphylococcus aureus involves binding of SP-A to the staphylococcal adhesin Eap and the macrophage receptors SP-A receptor 210 and scavenger receptor class A,” Journal of Biological Chemistry, vol. 286, no. 6, pp. 4854–4870, 2011. B. Sinha, P. P. Franc¸ois, O. N¨uße et al., “Fibronectin-binding protein acts as Staphylococcus aureus invasin via fibronectin bridging to integrin α5β1,” Cellular Microbiology, vol. 1, no. 2, pp. 101–117, 1999. S. Clement et al., “Evidence of an intracellular reservoir in the nasal mucosa of patients with recurrent Staphylococcus aureus rinosinusitis,” Journal of Infectious Diseases, vol. 192, pp. 1023–1028, 2005. C. Garzoni, P. Francois, A. Huyghe et al., “A global view of Staphylococcus aureus whole genome expression upon

[120]

[121]

[122]

[123]

[124]

[125]

[126]

[127]

[128]

[129]

[130]

[131]

[132]

[133]

internalization in human epithelial cells,” BMC Genomics, vol. 8, article 171, 2007. K. W. Bayles, C. A. Wesson, L. E. Liou, L. K. Fox, G. A. Bohach, and W. R. Trumble, “Intracellular Staphylococcus aureus escapes the endosome and induces apoptosis in epithelial cells,” Infection and Immunity, vol. 66, no. 1, pp. 336–342, 1998. C. A. Wesson, L. E. Liou, K. M. Todd, G. A. Bohach, W. R. Trumble, and K. W. Bayles, “Staphylococcus aureus Agr and Sar global regulators influence internalization and induction of apoptosis,” Infection and Immunity, vol. 66, no. 11, pp. 5238–5243, 1998. K. Dziewanowska, J. M. Patti, C. F. Deobald, K. W. Bayles, W. R. Trumble, and G. A. Bohach, “Fibronectin binding protein and host cell tyrosine kinase are required for internalization of Staphylococcus aureus by epithelial cells,” Infection and Immunity, vol. 67, no. 9, pp. 4673–4678, 1999. F. Agerer, A. Michel, K. Ohlsen, and C. R. Hauck, “Integrinmediated invasion of Staphylococcus aureus into human cells requires Src family protein-tyrosine kinases,” Journal of Biological Chemistry, vol. 278, no. 43, pp. 42524–42531, 2003. T. Fowler, S. Johansson, K. K. Wary, and M. H¨oo¨ k, “Src kinase has a central role in vitro cellular internalization of Staphylococcus aureus,” Cellular Microbiology, vol. 5, no. 6, pp. 417–426, 2003. F. J. Martin, M. I. Gomez, D. M. Wetzel et al., “Staphylococcus aureus activates type I IFN signaling in mice and humans through the Xr repeated sequences of protein A,” Journal of Clinical Investigation, vol. 119, no. 7, pp. 1931–1939, 2009. J. B. Wardenburg, R. J. Patel, and O. Schneewind, “Surface proteins and exotoxins are required for the pathogenesis of Staphylococcus aureus pneumonia,” Infection and Immunity, vol. 75, no. 2, pp. 1040–1044, 2007. ´ M. I. Gomez, A. Lee, B. Reddy et al., “Staphylococcus aureus protein A induces airway epithelial inflammatory responses by activating TNFR1,” Nature Medicine, vol. 10, no. 8, pp. 842–848, 2004. ´ M. I. Gomez, M. O’Seaghdha, M. Magargee, T. J. Foster, and A. S. Prince, “Staphylococcus aureus protein A activates TNFR1 signaling through conserved IgG binding domains,” Journal of Biological Chemistry, vol. 281, no. 29, pp. 20190– 20196, 2006. M. Li, B. A. Diep, A. E. Villaruz et al., “Evolution of virulence in epidemic community-associated methicillinresistant Staphylococcus aureus,” Proceedings of the National Academy of Sciences of the United States of America, vol. 106, no. 14, pp. 5883–5888, 2009. A. Fromageau, F. B. Gilbert, G. Pr´evost, and P. Rainard, “Binding of the Staphylococcus aureus leucotoxin LukM to its leucocyte targets,” Microbial Pathogenesis, vol. 49, no. 6, pp. 354–362, 2010. A. L. DuMont, T. K. Nygaard, R. L. Watkins et al., “Characterization of a new cytotoxin that contributes to Staphylococcus aureus pathogenesis,” Molecular Microbiology, vol. 79, no. 3, pp. 814–825, 2011. F. Rose, G. Dahlem, B. Guthmann et al., “Mediator generation and signaling events in alveolar epithelial cells attacked by S. aureus α-toxin,” American Journal of Physiology, vol. 282, no. 2, pp. L207–L214, 2002. S. Eichstaedt, K. G¨abler, S. Below et al., “Effects of Staphylococcus aureus-hemolysin A on calcium signalling in immortalized human airway epithelial cells,” Cell Calcium, vol. 45, no. 2, pp. 165–176, 2009.

Journal of Pathogens [134] G. A. Wilke and J. B. Wardenburg, “Role of a disintegrin and metalloprotease 10 in Staphylococcus aureus α-hemolysin—mediated cellular injury,” Proceedings of the National Academy of Sciences of the United States of America, vol. 107, no. 30, pp. 13473–13478, 2010. [135] A. Hayashida, A. H. Bartlett, T. J. Foster, and P. W. Park, “Staphylococcus aureus beta-toxin induces lung injury through syndecan-1,” American Journal of Pathology, vol. 174, no. 2, pp. 509–518, 2009. [136] M. Labandeira-Rey, F. Couzon, S. Boisset et al., “Staphylococcus aureus Panton-Valentine leukocidin causes necrotizing pneumonia,” Science, vol. 315, no. 5815, pp. 1130–1133, 2007. [137] J. B. Wardenburg, A. M. Palazzolo-Ballance, M. Otto, O. Schneewind, and F. R. DeLeo, “Panton-Valentine leukocidin is not a virulence determinant in murine models of community-associated methicillin-resistant Staphylococcus aureus disease,” Journal of Infectious Diseases, vol. 198, no. 8, pp. 1166–1170, 2008. [138] B. A. Diep, L. Chan, P. Tattevin et al., “Polymorphonuclear leukocytes mediate Staphylococcus aureus Panton-Valentine leukocidin-induced lung inflammation and injury,” Proceedings of the National Academy of Sciences of the United States of America, vol. 107, no. 12, pp. 5587–5592, 2010. [139] B. L¨offler, M. Hussain, M. Grundmeier et al., “Staphylococcus aureus Panton-Valentine leukocidin is a very potent cytotoxic factor for human neutrophils,” PLoS Pathogens, vol. 6, no. 1, Article ID e1000715, 2010. [140] A. Zivkovic, O. Sharif, K. Stich et al., “TLR 2 and CD14 mediate innate immunity and lung inflammation to staphylococcal Panton-Valentine leukocidin in vivo,” Journal of Immunology, vol. 186, no. 3, pp. 1608–1617, 2011. [141] K. L. Gage and M. Y. Kosoy, “Natural history of plague: perspectives from more than a century of research,” Annual Review of Entomology, vol. 50, pp. 505–528, 2005. [142] W. W. Lathem, S. D. Crosby, V. L. Miller, and W. E. Goldman, “Progression of primary pneumonic plague: a mouse model of infection, pathology, and bacterial transcriptional activity,” Proceedings of the National Academy of Sciences of the United States of America, vol. 102, no. 49, pp. 17786–17791, 2005. [143] R. Pollitzer, Plague, World Health Organization, Geneva, Switzerland, 1954. [144] R. Rebeil, R. K. Ernst, B. B. Gowen, S. I. Miller, and B. J. Hinnebusch, “Variation in lipid A structure in the pathogenic yersiniae,” Molecular Microbiology, vol. 52, no. 5, pp. 1363– 1373, 2004. [145] R. Rebeil, R. K. Ernst, C. O. Jarrett, K. N. Adams, S. I. Miller, and B. J. Hinnebusch, “Characterization of late acyltransferase genes of Yersinia pestis and their role in temperaturedependent lipid A variation,” Journal of Bacteriology, vol. 188, no. 4, pp. 1381–1388, 2006. [146] S. W. Montminy, N. Khan, S. McGrath et al., “Virulence factors of Yersinia pestis are overcome by a strong lipopolysaccharide response,” Nature Immunology, vol. 7, no. 10, pp. 1066–1073, 2006. [147] K. Kawahara, H. Tsukano, H. Watanabe, B. Lindner, and M. Matsuura, “Modification of the structure and activity of lipid A in Yersinia pestis lipopolysaccharide by growth temperature,” Infection and Immunity, vol. 70, no. 8, pp. 4092–4098, 2002. [148] M. Matsuura, H. Takahashi, H. Watanabe, S. Saito, and K. Kawahara, “Immunomodulatory effects of Yersinia pestis

15

[149]

[150]

[151]

[152]

[153]

[154]

[155]

[156]

[157]

[158]

[159]

[160]

[161]

[162]

lipopolysaccharides on human macrophages,” Clinical and Vaccine Immunology, vol. 17, no. 1, pp. 49–55, 2010. S. W. Montminy, N. Khan, S. McGrath et al., “Virulence factors of Yersinia pestis are overcome by a strong lipopolysaccharide response,” Nature Immunology, vol. 7, no. 10, pp. 1066–1073, 2006. M. V. Telepnev, G. R. Klimpel, J. Haithcoat, Y. A. Knirel, A. P. Anisimov, and V. L. Motin, “Tetraacylated lipopolysaccharide of Yersinia pestis can inhibit multiple toll-like receptormediated signaling pathways in human dendritic cells,” Journal of Infectious Diseases, vol. 200, no. 11, pp. 1694–1702, 2009. I. Adkins, M. K¨oberle, S. Gr¨obner, E. Bohn, I. B. Autenrieth, and S. Borgmann, “Yersinia outer proteins E, H, P, and T differentially target the cytoskeleton and inhibit phagocytic capacity of dendritic cells,” International Journal of Medical Microbiology, vol. 297, no. 4, pp. 235–244, 2007. T. Bergsbaken, S. L. Fink, and B. T. Cookson, “Pyroptosis: host cell death and inflammation,” Nature Reviews Microbiology, vol. 7, no. 2, pp. 99–109, 2009. J. L. Spinner, J. A. Cundiff, and S. D. Kobayashi, “Yersinia pestis type III secretion system-dependent inhibition of human polymorphonuclear leukocyte function,” Infection and Immunity, vol. 76, no. 8, pp. 3754–3760, 2008. C. M. Bosio, A. W. Goodyear, and S. W. Dow, “Early interaction of Yersinia pestis with APCs in the lung,” Journal of Immunology, vol. 175, no. 10, pp. 6750–6756, 2005. A. M. Cantwell, S. S. Bubeck, and P. H. Dube, “YopH inhibits early pro-inflammatory cytokine responses during plague pneumonia,” BMC Immunology, vol. 11, article 29, 2010. L. Zhou, A. Tan, and M. B. Hershenson, “Yersinia YopJ inhibits pro-inflammatory molecule expression in human bronchial epithelial cells,” Respiratory Physiology and Neurobiology, vol. 140, no. 1, pp. 89–97, 2004. N. Lemaˆıtre, F. Sebbane, D. Long, and B. J. Hinnebusch, “Yersinia pestis YopJ suppresses tumor necrosis factor alpha induction and contributes to apoptosis of immune cells in the lymph node but is not required for virulence in a rat model of bubonic plague,” Infection and Immunity, vol. 74, no. 9, pp. 5126–5131, 2006. T. M. Tsang, S. Felek, and E. S. Krukonis, “Ail binding to fibronectin facilitates Yersinia pestis binding to host cells and yop delivery,” Infection and Immunity, vol. 78, no. 8, pp. 3358–3368, 2010. K. L¨ahteenm¨aki, R. Virkola, A. Sar´en, L. Em¨ody, and T. K. Korhonen, “Expression of plasminogen activator Pla of Yersinia pestis enhances bacterial attachment to the mammalian extracellular matrix,” Infection and Immunity, vol. 66, no. 12, pp. 5755–5762, 1998. O. A. Sodeinde, Y. V. B. K. Subrahmanyam, K. Stark, T. Quan, Y. Bao, and J. D. Goguen, “A surface protease and the invasive character of plague,” Science, vol. 258, no. 5084, pp. 1004– 1007, 1992. Y. Zhang, J. Murtha, M. A. Roberts, R. M. Siegel, and J. B. Bliska, “Type III secretion decreases bacterial and host survival following phagocytosis of Yersinia pseudotuberculosis by macrophages,” Infection and Immunity, vol. 76, no. 9, pp. 4299–4310, 2008. E. M. Galv´an, H. Chen, and D. M. Schifferli, “The Psa fimbriae of Yersinia pestis interact with phosphatidylcholine on alveolar epithelial cells and pulmonary surfactant,” Infection and Immunity, vol. 75, no. 3, pp. 1272–1279, 2007.

16 [163] D. Payne, D. Tatham, E. D. Williamson, and R. W. Titball, “The pH 6 antigen of Yersinia pestis binds to β1-linked galactosyl residues in glycosphingolipids,” Infection and Immunity, vol. 66, no. 9, pp. 4545–4548, 1998. [164] F. Liu, H. Chen, E. M. Galv´an, M. A. Lasaro, and D. M. Schifferli, “Effects of Psa and F1 on the adhesive and invasive interactions of Yersinia pestis with human respiratory tract epithelial cells,” Infection and Immunity, vol. 74, no. 10, pp. 5636–5644, 2006. [165] E. H. Weening, J. S. Cathelyn, G. Kaufman et al., “The dependence of the Yersinia pestis capsule on pathogenesis is influenced by the mouse background,” Infection and Immunity, vol. 79, no. 2, pp. 644–652, 2011. [166] S. Felek, T. M. Tsang, and E. S. Krukonis, “Three Yersinia pestis adhesins facilitate Yop delivery to eukaryotic cells and contribute to plague virulence,” Infection and Immunity, vol. 78, no. 10, pp. 4134–4150, 2010. [167] A. M. Kolodziejek, D. R. Schnider, H. N. Rohde et al., “Outer membrane protein X (Ail) contributes to Yersinia pestis virulence in pneumonic plague and its activity is dependent on the lipopolysaccharide core length,” Infection and Immunity, vol. 78, no. 12, pp. 5233–5243, 2010. [168] W. W. Lathem, P. A. Price, V. L. Miller, and W. E. Goldman, “A plasminogen-activating protease specifically controls the development of primary pneumonic plague,” Science, vol. 315, no. 5811, pp. 509–513, 2007. [169] S. S. Bartra, K. L. Styer, D. M. O’Bryant et al., “Resistance of Yersinia pestis to complement-dependent killing is mediated by the ail outer membrane protein,” Infection and Immunity, vol. 76, no. 2, pp. 612–622, 2008. [170] A. M. Kolodziejek, D. R. Schnider, H. N. Rohde et al., “Outer membrane protein X (Ail) contributes to Yersinia pestis virulence in pneumonic plague and its activity is dependent on the lipopolysaccharide core length,” Infection and Immunity, vol. 78, no. 12, pp. 5233–5243, 2010. [171] E. M. Galv´an, M. A. S. Lasaro, and D. M. Schifferli, “Capsular antigen fraction 1 and Pla modulate the susceptibility of Yersinia pestis to pulmonary antimicrobial peptides such as cathelicidin,” Infection and Immunity, vol. 76, no. 4, pp. 1456–1464, 2008. [172] R. R. Isberg and J. M. Leong, “Multiple β1 chain integrins are receptors for invasin, a protein that promotes bacterial penetration into mammalian cells,” Cell, vol. 60, no. 5, pp. 861–871, 1990. [173] M. Simonet, B. Riot, N. Fortineau, and P. Berche, “Invasin production by Yersinia pestis is abolished by insertion of an IS200-like element within the inv gene,” Infection and Immunity, vol. 64, no. 1, pp. 375–379, 1996. [174] C. Pujol and J. B. Bliska, “The ability to replicate in macrophages is conserved between Yersinia pestis and Yersinia pseudotuberculosis,” Infection and Immunity, vol. 71, no. 10, pp. 5892–5899, 2003. [175] C. Pujol, J. P. Grabenstein, R. D. Perry, and J. B. Bliska, “Replication of Yersinia pestis in interferon γ-activated macrophages requires ripA, a gene encoded in the pigmentation locus,” Proceedings of the National Academy of Sciences of the United States of America, vol. 102, no. 36, pp. 12909–12914, 2005. [176] J. L. O’Loughlin, J. L. Spinner, S. A. Minnich, and S. D. Kobayashi, “Yersinia pestis two-component gene regulatory systems promote survival in human neutrophils,” Infection and Immunity, vol. 78, no. 2, pp. 733–782, 2010. [177] C. Pujol, K. A. Klein, G. A. Romanov et al., “Yersinia pestis can reside in autophagosomes and avoid xenophagy in murine

Journal of Pathogens

[178]

[179]

[180]

[181]

[182]

[183]

[184]

[185]

[186]

macrophages by preventing vacuole acidification,” Infection and Immunity, vol. 77, no. 6, pp. 2251–2261, 2009. J. D. Goguen, J. Yother, and S. C. Straley, “Genetic analysis of the low calcium response in Yersinia pestis Mu d1(Ap lac) insertion mutants,” Journal of Bacteriology, vol. 160, no. 3, pp. 842–848, 1984. J. P. Grabenstein, M. Marceau, C. Pujol, M. Simonet, and J. B. Bliska, “The response regulator PhoP of Yersinia pseudotuberculosis is important for replication in macrophages and for virulence,” Infection and Immunity, vol. 72, no. 9, pp. 4973– 4984, 2004. J. P. Grabenstein, H. S. Fukuto, L. E. Palmer, and J. B. Bliska, “Characterization of phagosome trafficking and identification of PhoP-regulated genes important for survival of Yersinia pestis in macrophages,” Infection and Immunity, vol. 74, no. 7, pp. 3727–3741, 2006. J. L. O’Loughlin, J. L. Spinner, S. A. Minnich, and S. D. Kobayashi, “Yersinia pestis two-component gene regulatory systems promote survival in human neutrophils,” Infection and Immunity, vol. 78, no. 2, pp. 733–782, 2010. N. A. Eisele and D. M. Anderson, “Dual-function antibodies to Yersinia pestis LcrV required for pulmonary clearance of plague,” Clinical and Vaccine Immunology, vol. 16, no. 12, pp. 1720–1727, 2009. B. L. Noel, S. Lilo, D. Capurso, J. Hill, and J. B. Bliska, “Yersinia pestis can bypass protective antibodies to LcrV and activation with gamma interferon to survive and induce apoptosis in murine macrophages,” Clinical and Vaccine Immunology, vol. 16, no. 10, pp. 1457–1466, 2009. N. A. Eisele, H. Lee-Lewis, C. Besch-Williford, C. R. Brown, and D. M. Anderson, “Chemokine receptor CXCR2 mediates bacterial clearance rather than neutrophil recruitment in a murine model of pneumonic plague,” American Journal of Pathology, vol. 178, no. 3, pp. 1190–1200, 2011. J. L. Spinner, K. S. Seo, J. L. O’Loughlin et al., “Neutrophils are resistant to Yersinia YopJ/P-induced apoptosis and are protected from ROS-mediated cell death by the type III secretion system,” PLoS ONE, vol. 5, no. 2, Article ID e9279, 2010. F. R. DeLeo, “Modulation of phagocyte apoptosis by bacterial pathogens,” Apoptosis, vol. 9, no. 4, pp. 399–413, 2004.